Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Nutrients ; 16(13)2024 Jun 21.
Article in English | MEDLINE | ID: mdl-38999736

ABSTRACT

The etiology and mechanisms of autism and autism spectrum disorder (ASD) are not yet fully understood. There is currently no treatment for ASD for providing significant improvement in core symptoms. Recent studies suggest, however, that ASD is associated with gut dysbiosis, indicating that modulation of gut microbiota in children with ASD may thus reduce the manifestation of ASD symptoms. The aim of this pilot study (prospective randomized, double-blinded, placebo-controlled) was to evaluate efficacy of the biological response modifier Juvenil in modulating the microbiome of children with ASD and, in particular, whether Juvenil is able to alleviate the symptoms of ASD. In total, 20 children with ASD and 12 neurotypical children were included in our study. Supplementation of ASD children lasted for three months. To confirm Juvenil's impact on the gut microbiome, stool samples were collected from all children and the microbiome's composition was analyzed. This pilot study demonstrated that the gut microbiome of ASD children differed significantly from that of healthy controls and was converted by Juvenil supplementation toward a more neurotypical microbiome that positively modulated children's autism symptoms.


Subject(s)
Autism Spectrum Disorder , Dietary Supplements , Gastrointestinal Microbiome , Humans , Pilot Projects , Double-Blind Method , Male , Female , Autism Spectrum Disorder/microbiology , Child , Feces/microbiology , Child, Preschool , Prospective Studies , Autistic Disorder/microbiology , Dysbiosis/microbiology
2.
Biology (Basel) ; 12(4)2023 Mar 25.
Article in English | MEDLINE | ID: mdl-37106700

ABSTRACT

Engagement of PRRs in recognition of PAMPs or DAMPs is one of the processes that initiates cellular stress. These sensors are involved in signaling pathways leading to induction of innate immune processes. Signaling initiated by PRRs is associated with the activation of MyD88-dependent signaling pathways and myddosome formation. MyD88 downstream signaling depends upon the context of signaling initiation, the cell (sub)type and the microenvironment of signal initiation. Recognition of PAMPs or DAMPs through PRRs activates the cellular autonomous defence mechanism, which orchestrates the cell responses to resolve specific insults at the single cell level. In general, stressed endoplasmic reticulum is directly linked with the induction of autophagy and initiation of mitochondrial stress. These processes are regulated by the release of Ca2+ from ER stores accepted by mitochondria, which respond through membrane depolarization and the production of reactive oxygen species generating signals leading to inflammasome activation. In parallel, signaling from PRRs initiates the accumulation of misfolded or inappropriately post-translationally modified proteins in the ER and triggers a group of conserved emergency rescue pathways known as unfolded protein response. The cell-autonomous effector mechanisms have evolutionarily ancient roots and were gradually specialized for the defence of specific cell (sub)types. All of these processes are common to the innate immune recognition of microbial pathogens and tumorigenesis as well. PRRs are active in both cases. Downstream are activated signaling pathways initiated by myddosomes, translated by the cellular autonomous defence mechanism, and finalized by inflammasomes.

3.
Microorganisms ; 9(10)2021 Oct 12.
Article in English | MEDLINE | ID: mdl-34683457

ABSTRACT

Immune responses to intracellular pathogens depend largely upon the activation of T helper type 1-dependent mechanisms. The contribution of B cells to establishing protective immunity has long been underestimated. Francisella tularensis, including a number of subspecies, provides a suitable model for the study of immune responses against intracellular bacterial pathogens. We previously demonstrated that Francisella infects B cells and activates B-cell subtypes to produce a number of cytokines and express the activation markers. Recently, we documented the early production of natural antibodies as a consequence of Francisella infection in mice. Here, we summarize current knowledge on the innate and acquired humoral immune responses initiated by Francisella infection and their relationships with the immune defense systems.

4.
Pathogens ; 10(3)2021 Mar 22.
Article in English | MEDLINE | ID: mdl-33809954

ABSTRACT

SARS-CoV-2 infection induces the production of autoantibodies, which is significantly associated with complications during hospitalization and a more severe prognosis in COVID-19 patients. Such a response of the patient's immune system may reflect (1) the dysregulation of the immune response or (2) it may be an attempt to regulate itself in situations where the non-infectious self poses a greater threat than the infectious non-self. Of significance may be the primary virus-host cell interaction where the surface-bound ACE2 ectoenzyme plays a critical role. Here, we present a brief analysis of recent findings concerning the immune recognition of SARS-CoV-2, which, we believe, favors the second possibility as the underlying reason for the production of autoantibodies during COVID-19.

5.
Sci Rep ; 11(1): 1541, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33452341

ABSTRACT

There remains to this day a great gap in understanding as to the role of B cells and their products-antibodies and cytokines-in mediating the protective response to Francisella tularensis, a Gram-negative coccobacillus belonging to the group of facultative intracellular bacterial pathogens. We previously have demonstrated that Francisella interacts directly with peritoneal B-1a cells. Here, we demonstrate that, as early as 12 h postinfection, germ-free mice infected with Francisella tularensis produce infection-induced antibody clones reacting with Francisella tularensis proteins having orthologs or analogs in eukaryotic cells. Production of some individual clones was limited in time and was influenced by virulence of the Francisella strain used. The phylogenetically stabilized defense mechanism can utilize these early infection-induced antibodies both to recognize components of the invading pathogens and to eliminate molecular residues of infection-damaged self cells.


Subject(s)
B-Lymphocytes/metabolism , Tularemia/immunology , Animals , Antibody Formation , B-Lymphocytes/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Francisella tularensis/pathogenicity , Mice , Mice, Inbred BALB C , Tularemia/microbiology , Virulence
6.
Microb Pathog ; 140: 103937, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31862393

ABSTRACT

Mycobacterium tuberculosis is the main etiological agent of tuberculosis. The Bacillus Calmette-Guérin (BCG) microbes that are primarily used as a vaccine against tuberculosis also constitute the dominant infection model for studying the interaction of mycobacteria with the host cell types. The majority of interaction experiments have been conducted using macrophages and monocytes as prototype phagocyte cell types. Here, we report that M. bovis BCG infects mouse primary B cells as well as human B cell line. The complement receptors, along with B cell receptors, are engaged in the process of bacterial entry into the host B cells. Once inside the B cells, the intracellular trafficking of BCG follows the complete endocytic pathway of the ingested particles, which is in contrast to the events taking place during ingestion of BCG by macrophages. In vivo infection of mice with M. bovis BCG activated peritoneal as well as splenic B cells to produce proinflammatory cytokines. This paper further supports the evidence that B cells are involved in a host's early interactions with intracellular bacterial pathogens and participate in the induction of innate defense responses.


Subject(s)
B-Lymphocytes , Cytokines/metabolism , Mycobacterium bovis/immunology , Tuberculosis/immunology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/microbiology , BCG Vaccine , Humans , Immunity, Innate , Mice , Primary Cell Culture , Tuberculosis/microbiology
7.
Article in English | MEDLINE | ID: mdl-31334134

ABSTRACT

Primary interaction of an intracellular bacterium with its host cell is initiated by activation of multiple signaling pathways in response to bacterium recognition itself or as cellular responses to stress induced by the bacterium. The leading molecules in these processes are cell surface membrane receptors as well as cytosolic pattern recognition receptors recognizing pathogen-associated molecular patterns or damage-associated molecular patterns induced by the invading bacterium. In this review, we demonstrate possible sequences of events leading to recognition of Francisella tularensis, present findings on known mechanisms for manipulating cell responses to protect Francisella from being killed, and discuss newly published data from the perspective of early stages of host-pathogen interaction.


Subject(s)
Francisella tularensis/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate , Pathogen-Associated Molecular Pattern Molecules/immunology , Receptors, Pattern Recognition/immunology , Tularemia/immunology , Alarmins/genetics , Alarmins/immunology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Francisella tularensis/genetics , Francisella tularensis/pathogenicity , Gene Expression Regulation , Host-Pathogen Interactions/genetics , Humans , Macrophages/immunology , Macrophages/microbiology , Pathogen-Associated Molecular Pattern Molecules/metabolism , Phagocytosis/genetics , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Pattern Recognition/genetics , Signal Transduction , Tularemia/genetics , Tularemia/microbiology
8.
Microb Pathog ; 123: 314-322, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30055244

ABSTRACT

Bacteria that are highly virulent, expressing high infectivity, and able to survive nebulization, pose great risk to the human population. One of these is Francisella tularensis, the etiological agent of tularemia. F. tularensis is a subject of intense scientific interest due to the fact that vaccines for its immunoprophylaxis in humans are not yet routinely available. One of the substantial obstacles in developing such vaccines is our insufficient knowledge of processes that initiate and regulate the expression of effective protective immunity against intracellular bacteria. Here, we present data documenting the different pattern of cellular behavior occurring in an environment unaffected by microbiota using the model of germ-free mice mono-associated with F. tularensis subsp. holarctica strain LVS in comparison with a classic specific-pathogen-free murine model during early stages of infection.


Subject(s)
Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Host-Pathogen Interactions/immunology , Tularemia/immunology , Animals , Bacterial Vaccines/immunology , Cytokines/metabolism , Disease Models, Animal , Female , Germ-Free Life/immunology , Immunity, Innate , Mice , Mice, Inbred BALB C , Microbiota , Peritoneum/microbiology , Peritoneum/pathology , Specific Pathogen-Free Organisms/immunology , Spleen/microbiology , Spleen/pathology , Tularemia/microbiology , Tularemia/pathology
9.
Article in English | MEDLINE | ID: mdl-29085810

ABSTRACT

The intracellular bacterial pathogen Francisella tularensis causes serious infectious disease in humans and animals. Moreover, F. tularensis, a highly infectious pathogen, poses a major concern for the public as a bacterium classified under Category A of bioterrorism agents. Unfortunately, research has so far failed to develop effective vaccines, due in part to the fact that the pathogenesis of intracellular bacteria is not fully understood and in part to gaps in our understanding of innate immune recognition processes leading to the induction of adaptive immune response. Recent evidence supports the concept that immune response to external stimuli in the form of bacteria is guided by the primary interaction of the bacterium with the host cell. Based on data from different Francisella models, we present here the basic paradigms of the emerging innate immune recognition concept. According to this concept, the type of cell and its receptor(s) that initially interact with the target constitute the first signaling window; the signals produced in the course of primary interaction of the target with a reacting cell act in a paracrine manner; and the innate immune recognition process as a whole consists in a series of signaling windows modulating adaptive immune response. Finally, the host, in the strict sense, is the interacting cell.


Subject(s)
Francisella tularensis/immunology , Host-Pathogen Interactions/immunology , Immunity, Innate , Tularemia/immunology , Adaptive Immunity , Animals , Disease Models, Animal , Humans , Immune System , Mice , Paracrine Communication/immunology
10.
Cell Mol Life Sci ; 73(20): 3961-9, 2016 10.
Article in English | MEDLINE | ID: mdl-27544211

ABSTRACT

This brief review is dedicated to the legacy of Prof. Jaroslav Sterzl and his colleagues, who laid the foundation for gnotobiology in the former Czechoslovakia 55 years. Prof. Sterzl became one of the founders of modern Czechoslovak immunology, which was characterized by work on a wide range of problems needing to be solved. While examining the mechanisms of innate immunity, he focused his studies on the induction of antibody production by immunocompetent cells involved in adaptive immune transmission while using the model of pig fetuses and germ-free piglets and characterizing immunoglobulins in the sera of these piglets. Although not fully appreciated to this day, his experimental proof of the hypothesis focused on the common precursor of cell-forming antibodies of different isotypes was later confirmed in experiments at the gene level. Prof. Sterzl's work represented a true milestone in the development of not solely Czechoslovak but also European and global immunology. He collaborated closely with the World Health Organization for many years, serving there as leader of the Reference Laboratory for Factors of Innate Immunity.


Subject(s)
Germ-Free Life , Host-Pathogen Interactions , Animals , Disease Models, Animal , Gastrointestinal Microbiome , Mice
11.
PLoS One ; 10(7): e0132571, 2015.
Article in English | MEDLINE | ID: mdl-26161475

ABSTRACT

Francisella tularensis, the etiological agent of tularemia, is an intracellular pathogen that dominantly infects and proliferates inside phagocytic cells but can be seen also in non-phagocytic cells, including B cells. Although protective immunity is known to be almost exclusively associated with the type 1 pathway of cellular immunity, a significant role of B cells in immune responses already has been demonstrated. Whether their role is associated with antibody-dependent or antibody-independent B cell functions is not yet fully understood. The character of early events during B cell-pathogen interaction may determine the type of B cell response regulating the induction of adaptive immunity. We used fluorescence microscopy and flow cytometry to identify the basic requirements for the entry of F. tularensis into B cells within in vivo and in vitro infection models. Here, we present data showing that Francisella tularensis subsp. holarctica strain LVS significantly infects individual subsets of murine peritoneal B cells early after infection. Depending on a given B cell subset, uptake of Francisella into B cells is mediated by B cell receptors (BCRs) with or without complement receptor CR1/2. However, F. tularensis strain FSC200 ΔiglC and ΔftdsbA deletion mutants are defective in the ability to enter B cells. Once internalized into B cells, F. tularensis LVS intracellular trafficking occurs along the endosomal pathway, albeit without significant multiplication. The results strongly suggest that BCRs alone within the B-1a subset can ensure the internalization process while the BCRs on B-1b and B-2 cells need co-signaling from the co receptor containing CR1/2 to initiate F. tularensis engulfment. In this case, fluidity of the surface cell membrane is a prerequisite for the bacteria's internalization. The results substantially underline the functional heterogeneity of B cell subsets in relation to F. tularensis.


Subject(s)
B-Lymphocytes/metabolism , Francisella tularensis/physiology , Receptors, Antigen, B-Cell/metabolism , Receptors, Complement/metabolism , Tularemia/microbiology , Animals , B-Lymphocytes/microbiology , Biological Transport , Cell Line , Female , Genes, Bacterial , Host-Pathogen Interactions , Membrane Microdomains/metabolism , Mice, Inbred BALB C , Microbial Viability , Receptors, IgG/metabolism , Sequence Deletion
12.
Microb Pathog ; 75: 49-58, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25200734

ABSTRACT

Francisella tularensis, a facultative intracellular Gram-negative bacterium, causes the illness tularemia. The infection of mice with live vaccine strain is considered to be a model of human tularemia. F. tularensis infects predominantly such phagocytic cells as macrophages or neutrophils, but it also infects non-phagocytic hepatocytes, epithelial cells, and murine and human B cell lines. Based on work with the murine tularemia model, we report here that F. tularensis LVS infects peritoneal CD19(+) cells - exclusively B-1a cells - early after intraperitoneal infection in vivo. The peritoneal and consequently spleen CD19(+) cells are activated by the F. tularensis LVS infection to express the activation markers from MHC class II, CD25, CD54, CD69, and the co-stimulatory molecules CD80 and CD86. As early as 12 h post-infection, the peritoneal CD19(+) cells produce IFN-γ, IL-1ß, IL-4, IL-6, IL-12, IL-17, IL-23, and TNF-α. The spleen CD19(+) cells respond to infection with some delay. Moreover, the F. tularensis infected A20 B cell line activates CD3(+) spleen cells isolated from naïve mice. Thus, the data presented here suggest that B cells have all the attributes to actively participate in the induction and regulation of the adaptive immune response during early stages of F. tularensis infection.


Subject(s)
B-Lymphocyte Subsets/immunology , Cytokines/metabolism , Lymphocyte Activation , Tularemia/immunology , Animals , Antigens, CD/analysis , B-Lymphocyte Subsets/chemistry , Disease Models, Animal , Female , Histocompatibility Antigens Class II/analysis , Mice, Inbred BALB C , Peritoneum/immunology , Spleen/immunology , Time Factors
13.
Appl Microbiol Biotechnol ; 97(23): 10103-15, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24162084

ABSTRACT

The intracellular pathogens have the unique capacity to sense the host cell environment and to respond to it by alteration in gene expression and protein synthesis. Proteomic analysis of bacteria exposed directly to the host cell milieu might thus greatly contribute to the elucidation of processes leading to bacterial adaptation and proliferation inside the host cell. Here we have performed a global proteome analysis of a virulent Francisella tularensis subsp. holarctica strain during its intracellular cycle within the macrophage-like murine cell line J774.2 using the metabolic pulse-labeling of bacterial proteins with (35)S-methionine and (35)S-cysteine in various periods of infection. The two-dimensional gel analysis revealed macrophage-induced bacterial proteome changes in which 64 identified proteins were differentially expressed in comparison to controls grown in tissue culture medium. Nevertheless, activation of macrophages with interferon gamma before in vitro infection decreased the number of detected alterations in protein levels. Thus, these proteomic data indicate the F. tularensis ability to adapt to the intracellular hostile environment that is, however, diminished by prior interferon gamma treatment of host cells.


Subject(s)
Bacterial Proteins/chemistry , Francisella tularensis/physiology , Host-Pathogen Interactions , Tularemia/microbiology , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Electrophoresis, Gel, Two-Dimensional , Francisella tularensis/chemistry , Francisella tularensis/genetics , Humans , Macrophages/microbiology , Mice , Molecular Sequence Data , Proteomics
14.
Microb Pathog ; 53(5-6): 259-68, 2012.
Article in English | MEDLINE | ID: mdl-22841607

ABSTRACT

The role of antibodies in the course of Francisella tularensis (F. tularensis) infection is still a subject of debate. The understanding of the poorly described role of humoral immunity is more than important for the effort to develop effective prophylactic procedure against the infection with Francisella virulent strains. We utilized the model of gamma-irradiated mice for the studies of the protective role of anti-F. tularensis antibodies in order to partially eliminate cellular responses. The model of gamma-irradiated mice can also demonstrate the responses of immunocompromised host to intracellular bacterial infection. The gamma-irradiation by doses greater than 3 Gy completely impairs the resistance to infection and causes a disbalance of cytokine production in mice. In this study, we demonstrate that passive transfer of immune sera protected irradiated mice against subsequent infection with strains of F. tularensis subsp. holarctica. Naïve mice of BALB/c or C3H/CBi strains were subjected to passive transfer of sera obtained from immunized mice with live vaccine strain (LVS) F. tularensis LVS, F. tularensis subsp. holarctica strain 15, heat-killed F. tularensis LVS, or heat-killed strain 15 two hours before infection with lethal doses of LVS or strain 15. The passive transfer of sera obtained from immunized mice conferred full protection of naïve unirradiated as well as sublethally irradiated mice against low lethal doses of infection with F. tularensis LVS or strain 15, in all variants of the experiments. In addition, the passively protected mice that survived the primary infection with F. tularensis LVS were protected also against further secondary challenge with a highly virulent strain of F. tularensis subsp. tularensis SchuS4. Moreover, the first evidence of combination of successful passive transfer of immunity by specific antisera and subsequent active immunization of immunocompromised animals is demonstrated. In summary, we demonstrate that B cell-mediated effector responses together with the induction of T cell-mediated immunity both play an important role in naïve and also in immunocompromised mice and this fact it would be appropriate to take into the account in the design of new vaccines.


Subject(s)
Antibodies, Bacterial/blood , Francisella tularensis/immunology , Francisella tularensis/pathogenicity , Tularemia/prevention & control , Animals , Disease Models, Animal , Female , Gamma Rays , Humans , Immunization, Passive , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Survival Analysis
15.
Proteomics ; 10(24): 4501-11, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21136602

ABSTRACT

The facultative intracellular bacterium Francisella tularensis is the causal agent of the serious infectious disease tularemia. Despite the dynamic progress, which has been made in last few years, important questions regarding Francisella pathogenicity still remain to be answered. Generally, secreted proteins play an important role in pathogenicity of intracellular microbes. In this study, we investigated the protein composition of the culture filtrate proteins of highly virulent F. tularensis subsp. tularensis, strain SCHU S4 and attenuated F. tularensis subsp. holarctica, live vaccine strain using a comparative proteomic analysis. The majority of proteins identified in this study have been implicated in virulence mechanisms of other pathogens, and several have been categorized as having moonlighting properties; those that have more than one unrelated function. This profiling study of secreted proteins resulted in the unique detection of acid phosphatase (precursor) A (AcpA), ß-lactamase, and hypothetical protein FTT0484 in the highly virulent strain SCHU S4 secretome. The release of AcpA may be of importance for F. tularensis subsp. tularensis virulence due to the recently described AcpA role in the F. tularensis escape from phagosomes.


Subject(s)
Bacterial Proteins/chemistry , Francisella tularensis/chemistry , Proteome/chemistry , Culture Media, Conditioned , Francisella tularensis/pathogenicity , Virulence Factors/chemistry
16.
Microb Pathog ; 49(5): 226-36, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20600796

ABSTRACT

Francisella tularensis is a facultative intracellular, gram-negative bacterium that induces apoptosis in macrophages and B cells. Here we show apoptotic pathways that are activated in the Ramos human B cell line in the course of F. tularensis infection. Live bacteria F. tularensis FSC200 activate caspases 8, 9 and 3, as well as Bid; release cytochrome c and apoptosis-inducing factor from mitochondria; and induce depolarization of mitochondrial membrane potential in the Ramos cell line, thus leading these cells to apoptosis. Unlike live bacteria, killed F. tularensis FSC200 bacteria activated only caspase 3, and did not cause apoptosis of Ramos cells as measured by annexin V. Killed bacteria also caused accumulation of anti-apoptotic protein Bclx(L) in mitochondrial membranes. Thus, live F. tularensis activates both caspase pathways (receptor-mediated and intrinsic) as well as caspase-independent mitochondrial death.


Subject(s)
Apoptosis , B-Lymphocytes/microbiology , Francisella tularensis/pathogenicity , BH3 Interacting Domain Death Agonist Protein/biosynthesis , Caspase 3/biosynthesis , Caspase 8/biosynthesis , Caspase 9/biosynthesis , Cell Line , Cytochromes c/metabolism , Humans , Membrane Potential, Mitochondrial , Mitochondria/enzymology , Mitochondria/physiology
17.
Anal Chem ; 80(18): 7097-104, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18707130

ABSTRACT

Rapid and reliable detection, identification, and typing of bacterial species are necessary in response to natural or terrorist-caused outbreaks of infectious diseases and play crucial roles in diagnosis and efficient treatment. We report here two proteomic approaches with a high potential in the detection and identification of Coxiella burnetii, the causative agent of Q fever. The first of them starts with the acetonitrile (ACN) and trichloroacetic acid extractions of inactivated C. burnetii cells followed by the detection of extracted molecules and ions derived from the inactivated cells by matrix-assisted laser desorption/ionization time-of-flight mass spectrometry. In the second approach, identification of the proteins extracted by ACN is accomplished after enzymatic digestion by electrospray tandem mass spectrometry coupled to a nanoscale ultraperformance liquid chromatography (LC-MS/MS). In order to observe morphological differences on the surface structures upon extraction, the inactivated and treated cells of the bacterium were examined by electron microscopy. The LC-MS/MS approach has allowed identification of 20 proteins in the ACN extracts of C. burnetii strain RSA 493 that were observed in more than 3 out of 10 experiments.


Subject(s)
Bacterial Proteins/analysis , Bacterial Proteins/isolation & purification , Coxiella burnetii/chemistry , Coxiella burnetii/isolation & purification , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Chromatography, Liquid , Databases, Factual , Microscopy, Electron , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry , Trypsin/metabolism
18.
Microb Pathog ; 45(2): 79-85, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18524531

ABSTRACT

Immunity to Francisella tularensis is largely mediated by T lymphocytes but an important role of B lymphocytes in early stage of infection was previously uncovered. We wanted to find out if F. tularensis is able to infect B cells and/or influence them by direct contact. To investigate this possibility we infected B cell lines from mouse (A20) or humans (Ramos RA-1), or primary mouse spleen cells, with F. tularensis LVS and F. tularensis FSC200 in vitro. In all cases, we detected bacteria on the cell surface and inside the B cells using transmission electron microscopy. More than 20% cells were infected by microbes after 24h. The number of bacteria, determined by CFU, increased about 1 log during 24h. Infection with live bacteria led to apoptosis of Ramos cells and mouse CD19(+) spleen cells. Approximately 30% of cells were apoptotic after 24h and 70% after 48 h, independently of the F. tularensis strain, while only 10% of non-infected cell were apoptotic at either time point. Apoptosis was confirmed by Western blot using anti-PARP antibodies. Thus, this study demonstrates unique phenomenon - namely, the ability of the intracellular pathogen F. tularensis to invade and induce apoptosis in B cells.


Subject(s)
Apoptosis , B-Lymphocytes/microbiology , Francisella tularensis/physiology , Animals , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Cell Line , Francisella tularensis/immunology , Humans , Mice , Microscopy, Electron, Transmission
19.
Immunol Lett ; 108(2): 151-9, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17241671

ABSTRACT

Francisella tularensis is a Gram-negative, facultative intracellular bacterium causing disease in many mammalian species. The low infectious dose of F. tularensis and the ease of air-borne transmission are the main features responsible for the classification of this bacterium as a potential biological weapon. The live attenuated strain of F. tularensis live vaccine strain (LVS) is currently only effective vaccine against tularemia, however, this type of vaccine has not been approved for human use. In the presented study, sub-immunoproteome analysis was performed to search for new immunogenic proteins of Francisella tularensis LVS grown under different conditions. By this approach 35 immunoreactive antigens were identified, 19 of them showed to be novel immunogens. In conclusion, sub-immunoproteome analysis resulted in successful identification of novel immunoreactive proteins.


Subject(s)
Antigens, Bacterial/analysis , Francisella tularensis/immunology , Membrane Proteins/immunology , Antigens, Bacterial/immunology , Antigens, Bacterial/isolation & purification , Computational Biology , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Humans , Immune Sera/immunology , Isoelectric Focusing , Membrane Proteins/analysis , Membrane Proteins/isolation & purification , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
20.
FEMS Microbiol Lett ; 269(1): 11-21, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17227466

ABSTRACT

Francisella tularensis is a highly virulent, facultative intracellular pathogen that causes tularemia in humans and animals. Although it is one of the most infectious bacterial pathogens, little is known about its virulence mechanisms. In this study, the response of F. tularensis live vaccine strain to iron depletion, which simulates the environment within the host, was investigated. In order to detect alterations in protein synthesis, metabolic labeling, followed by 2D-PAGE analysis was used. Globally, 141 protein spots were detected whose levels were significantly altered in the iron-restricted medium. About 65% of the spots were successfully identified using mass spectrometric approaches. Importantly, among the proteins produced at an increased level during iron-limited growth, three proteins were found encoded by the igl operon, located in the F. tularensis pathogenicity island I (FPI). Of these, the IglC and IglA proteins were previously reported to be necessary for full virulence of F. tularensis. These results, obtained at the proteome level, support and confirm recently published data showing that the igl operon genes are transcribed in response to iron limitation.


Subject(s)
Bacterial Proteins/metabolism , Francisella tularensis/metabolism , Francisella tularensis/pathogenicity , Genomic Islands , Iron/metabolism , Virulence Factors/metabolism , Bacterial Proteins/genetics , Electrophoresis, Gel, Two-Dimensional , Francisella tularensis/growth & development , Gene Expression Profiling , Gene Expression Regulation, Bacterial , Mass Spectrometry , Operon , Proteomics , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL